1
|
Aerobic Exercise Training Reduces Atherogenesis Induced by Low-Sodium Diet in LDL Receptor Knockout Mice. Antioxidants (Basel) 2022; 11:antiox11102023. [PMID: 36290746 PMCID: PMC9598599 DOI: 10.3390/antiox11102023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 11/24/2022] Open
Abstract
This study investigated the efficacy of aerobic exercise training (AET) in the prevention of dyslipidemia, insulin resistance (IR), and atherogenesis induced by severe low-sodium (LS) diet. LDL receptor knockout (LDLR KO) mice were fed a low-sodium (LS) (0.15% NaCl) or normal-sodium (NS; 1.27% NaCl) diet, submitted to AET in a treadmill, 5 times/week, 60 min/day, 15 m/min, for 90 days, or kept sedentary. Blood pressure (BP), plasma total cholesterol (TC) and triglyceride (TG) concentrations, lipoprotein profile, and insulin sensitivity were evaluated at the end of the AET protocol. Lipid infiltration, angiotensin II type 1 receptor (AT1), receptor for advanced glycation end products (RAGE), carboxymethyllysine (CML), and 4-hydroxynonenal (4-HNE) contents as well as gene expression were determined in the brachiocephalic trunk. BP and TC and gene expression were similar among groups. Compared to the NS diet, the LS diet increased vascular lipid infiltration, CML, RAGE, 4-HNE, plasma TG, LDL-cholesterol, and VLDL-TG. Conversely, the LS diet reduced vascular AT1 receptor, insulin sensitivity, HDL-cholesterol, and HDL-TG. AET prevented arterial lipid infiltration; increases in CML, RAGE, and 4-HNE contents; and reduced AT1 levels and improved LS-induced peripheral IR. The current study showed that AET counteracted the deleterious effects of chronic LS diet in an atherogenesis-prone model by ameliorating peripheral IR, lipid infiltration, CML, RAGE, 4-HNE, and AT1 receptor in the intima-media of the brachiocephalic trunk. These events occurred independently of the amelioration of plasma-lipid profile, which was negatively affected by the severe dietary-sodium restriction.
Collapse
|
2
|
Ding Q, Shao C, Rose P, Zhu YZ. Epigenetics and Vascular Senescence-Potential New Therapeutic Targets? Front Pharmacol 2020; 11:535395. [PMID: 33101015 PMCID: PMC7556287 DOI: 10.3389/fphar.2020.535395] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 08/31/2020] [Indexed: 12/14/2022] Open
Abstract
Epigenetics is defined as the heritable alterations of gene expression without changes to the coding sequence of DNA. These alterations are mediated by processes including DNA methylation, histone modifications, and non-coding RNAs mechanisms. Vascular aging consists of both structural and functional changes in the vasculature including pathological processes that drive progression such as vascular cell senescence, inflammation, oxidation stress, and calcification. As humans age, these pathological conditions gradually accumulate, driven by epigenetic alterations, and are linked to various aging-related diseases. The development of drugs targeting a spectrum of epigenetic processes therefore offers novel treatment strategies for the targeting of age-related diseases. In our previous studies, we identified HDAC4, JMJD3, Fra-1, and GATA4 as potential pharmacological targets for regulating vascular inflammation, injury, and senescence.
Collapse
Affiliation(s)
- Qian Ding
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China.,School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Chunhong Shao
- Department of Psychiatry, Huashan Hospital, Fudan University, Shanghai, China
| | - Peter Rose
- School of Biosciences, University of Nottingham, Loughborough, United Kingdom
| | - Yi Zhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China
| |
Collapse
|
3
|
Abstract
PURPOSE OF REVIEW To summarize all available data on targeting angiotensinogen with RNA-based therapeutics as a new tool to combat cardiovascular diseases. RECENT FINDINGS Liver-targeted, stable antisense oligonucleotides and small interfering RNA targeting angiotensinogen are now available, and may allow treatment with at most a few injections per year, thereby improving adherence. Promising results have been obtained in hypertensive animal models, as well as in rodent models of atherosclerosis, polycystic kidney disease and pulmonary fibrosis. The next step will be to evaluate the optimal degree of suppression, synergy with existing renin-angiotensin-aldosterone system blockers, and to determine harmful effects of suppressing angiotensinogen in the context of common comorbidities, such as heart failure and chronic kidney disease. SUMMARY Targeting angiotensinogen with RNA-based therapeutics is a promising new tool to treat hypertension and diseases beyond. Their long-lasting effects are particularly exciting, and if translated to a clinical application of at most a few administrations per year, may help to eliminate nonadherence.
Collapse
|
4
|
St. Paul A, Corbett CB, Okune R, Autieri MV. Angiotensin II, Hypercholesterolemia, and Vascular Smooth Muscle Cells: A Perfect Trio for Vascular Pathology. Int J Mol Sci 2020; 21:E4525. [PMID: 32630530 PMCID: PMC7350267 DOI: 10.3390/ijms21124525] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/16/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality in the Western and developing world, and the incidence of cardiovascular disease is increasing with the longer lifespan afforded by our modern lifestyle. Vascular diseases including coronary heart disease, high blood pressure, and stroke comprise the majority of cardiovascular diseases, and therefore represent a significant medical and socioeconomic burden on our society. It may not be surprising that these conditions overlap and potentiate each other when we consider the many cellular and molecular similarities between them. These intersecting points are manifested in clinical studies in which lipid lowering therapies reduce blood pressure, and anti-hypertensive medications reduce atherosclerotic plaque. At the molecular level, the vascular smooth muscle cell (VSMC) is the target, integrator, and effector cell of both atherogenic and the major effector protein of the hypertensive signal Angiotensin II (Ang II). Together, these signals can potentiate each other and prime the artery and exacerbate hypertension and atherosclerosis. Therefore, VSMCs are the fulcrum in progression of these diseases and, therefore, understanding the effects of atherogenic stimuli and Ang II on the VSMC is key to understanding and treating atherosclerosis and hypertension. In this review, we will examine studies in which hypertension and atherosclerosis intersect on the VSMC, and illustrate common pathways between these two diseases and vascular aging.
Collapse
Affiliation(s)
| | | | | | - Michael V. Autieri
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA; (A.S.P.); (C.B.C.); (R.O.)
| |
Collapse
|
5
|
Kukida M, Sawada H, Daugherty A, Lu HS. Megalin: A bridge connecting kidney, the renin-angiotensin system, and atherosclerosis. Pharmacol Res 2020; 151:104537. [PMID: 31707037 PMCID: PMC6980733 DOI: 10.1016/j.phrs.2019.104537] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 11/05/2019] [Indexed: 01/21/2023]
Abstract
Megalin is a member of the low-density lipoprotein receptor superfamily. It has been recognized as an endocytic receptor for a large spectrum of ligands. As a consequence, megalin regulates homeostasis of many molecules and affects multiple physiological and pathophysiological functions. The renin-angiotensin system is a hormonal system. A number of studies have reported contributions of the renin-angiotensin system to atherosclerosis. There is evolving evidence that megalin is a regulator of the renin-angiotensin system, and contributes to atherosclerosis. This brief review provides contemporary insights into effects of megalin on renal functions, the renin-angiotensin system, and atherosclerosis.
Collapse
Affiliation(s)
- Masayoshi Kukida
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Hisashi Sawada
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Hong S Lu
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
6
|
Antisense oligonucleotides targeting angiotensinogen: insights from animal studies. Biosci Rep 2019; 39:BSR20180201. [PMID: 30530571 PMCID: PMC6328882 DOI: 10.1042/bsr20180201] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/21/2018] [Accepted: 12/07/2018] [Indexed: 01/01/2023] Open
Abstract
Angiotensinogen (AGT) is the unique substrate of all angiotensin peptides. We review the recent preclinical research of AGT antisense oligonucleotides (ASOs), a rapidly evolving therapeutic approach. The scope of the research findings not only opens doors for potentially new therapeutics of hypertension and many other diseases, but also provides insights into understanding critical physiological and pathophysiological roles mediated by AGT.
Collapse
|
7
|
Bian F, Cui J, Zheng T, Jin S. Reactive oxygen species mediate angiotensin II-induced transcytosis of low-density lipoprotein across endothelial cells. Int J Mol Med 2017; 39:629-635. [PMID: 28204818 PMCID: PMC5360350 DOI: 10.3892/ijmm.2017.2887] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 01/03/2017] [Indexed: 11/06/2022] Open
Abstract
The retention of plasma low-density lipoprotein (LDL) particles to subendothelial spaces through transcytosis across the endothelium is the initial step of atherosclerosis (AS). Angiotensin II (Ang II), as the principal effector molecule of the renin-angiotensin system (RAS), is implicated in several important steps of AS development. However, whether or not Ang II can directly exert a pro‑atherogenic effect by promoting LDL transcytosis across endothelial barriers, has not been defined. In the present study, we found that Ang II upregulated intracellular reactive oxygen species (ROS) levels in endothelial cells (ECs) by measuring fluorescence of 2',7'-dichlorofluorescein (DCF‑DA). Based on our transcytosis model, we observed that Ang II significantly accelerated LDL transcytosis, whereas transcytosis inhibitor methyl-β-cyclodextrin (MβCD) and ROS inhibitor dithiothreitol (DTT), markedly blocked the Ang II-stimulated increase in LDL transcytosis. Confocal imaging analysis revealed that both LDL uptake by cells and LDL retention in human umbilical venous walls were highly elevated after Ang II exposure, while MβCD and DTT significantly inhibited the effects of Ang II. What is more, proteins involved in caveolae-mediated transcytosis, including LDL receptor (LDLR), caveolin-1 and cavin-1, were associated with Ang II-induced LDL transcytosis across the ECs. Nevertheless, this process was independent of clathrin in our study. Of note, ROS inhibitor, DTT, markedly decreased the expression levels of those proteins. Consequently, ROS are critical mediators in Ang II-induced LDL transcytosis. Hopefully, these findings will provide novel insight into the crosstalk between dyslipidemia and RAS in atherogenesis.
Collapse
Affiliation(s)
- Fang Bian
- Department of Pharmacy, The Affiliated Hospital of Xiangyang Central Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, P.R. China
| | - Jun Cui
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Xiangyang Central Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, P.R. China
| | - Tao Zheng
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Si Jin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
8
|
Sakamuri SSVP, Higashi Y, Sukhanov S, Siddesha JM, Delafontaine P, Siebenlist U, Chandrasekar B. TRAF3IP2 mediates atherosclerotic plaque development and vulnerability in ApoE(-/-) mice. Atherosclerosis 2016; 252:153-160. [PMID: 27237075 DOI: 10.1016/j.atherosclerosis.2016.05.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/12/2016] [Accepted: 05/18/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND AND AIMS Atherosclerosis is a major cause of heart attack and stroke. Inflammation plays a critical role in the development of atherosclerosis. Since the cytoplasmic adaptor molecule TRAF3IP2 (TRAF3-Interacting Protein 2) plays a causal role in various autoimmune and inflammatory diseases, we hypothesized that TRAF3IP2 mediates atherosclerotic plaque development. METHODS TRAF3IP2/ApoE double knockout (DKO) mice were generated by crossing TRAF3IP2(-/-) and ApoE(-/-) mice. ApoE(-/-) mice served as controls. Both DKO and control mice were fed a high-fat diet for 12 weeks. Plasma lipids were measured by ELISA, atherosclerosis by en face analysis of aorta and plaque cross-section measurements at the aortic valve region, plaque necrotic core area, collagen and smooth muscle cell (SMC) content by histomorphometry, and aortic gene expression by RT-qPCR. RESULTS The plasma lipoprotein profile was not altered by TRAF3IP2 gene deletion in ApoE(-/-) mice. While total aortic plaque area was decreased in DKO female, but not male mice, the plaque necrotic area was significantly decreased in DKO mice of both genders. Plaque collagen and SMC contents were increased significantly in both female and male DKO mice compared to respective controls. Aortic expression of proinflammatory cytokine (Tumor necrosis factor α, TNFα), chemokine (Chemokine (C-X-C motif) Ligand 1, CXCL1) and adhesion molecule (Vascular cell adhesion molecule 1, VCAM1; and Intercellular adhesion molecule 1, ICAM1) gene expression were decreased in both male and female DKO mice. In addition, the male DKO mice expressed markedly reduced levels of extracellular matrix (ECM)-related genes, including TIMP1 (Tissue inhibitor of metalloproteinase 1), RECK (Reversion-Inducing-Cysteine-Rich Protein with Kazal Motifs) and ADAM17 (A Disintegrin And Metalloproteinase 17). CONCLUSIONS TRAF3IP2 plays a causal role in atherosclerotic plaque development and vulnerability, possibly by inducing the expression of multiple proinflammatory mediators. TRAF3IP2 could be a potential therapeutic target in atherosclerotic vascular diseases.
Collapse
Affiliation(s)
| | - Yusuke Higashi
- Heart and Vascular Institute, Tulane University School of Medicine, New Orleans, LA, 70112, United States
| | - Sergiy Sukhanov
- Heart and Vascular Institute, Tulane University School of Medicine, New Orleans, LA, 70112, United States
| | - Jalahalli M Siddesha
- Heart and Vascular Institute, Tulane University School of Medicine, New Orleans, LA, 70112, United States
| | - Patrice Delafontaine
- Heart and Vascular Institute, Tulane University School of Medicine, New Orleans, LA, 70112, United States
| | - Ulrich Siebenlist
- Laboratory of Immunoregulation, NIAID/NIH, Bethesda, MD, 20892, United States
| | - Bysani Chandrasekar
- Heart and Vascular Institute, Tulane University School of Medicine, New Orleans, LA, 70112, United States; HS Truman Memorial Veterans Hospital, 800 Hospital Drive, Columbia, MO, 75201, United States.
| |
Collapse
|
9
|
Toro L, Li M, Zhang Z, Singh H, Wu Y, Stefani E. MaxiK channel and cell signalling. Pflugers Arch 2014; 466:875-86. [PMID: 24077696 DOI: 10.1007/s00424-013-1359-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 09/09/2013] [Accepted: 09/10/2013] [Indexed: 01/23/2023]
Abstract
The large-conductance Ca2+- and voltage-activated K+ (MaxiK, BK, BKCa, Slo1, KCa1.1) channel role in cell signalling is becoming apparent as we learn how the channel interacts with a multiplicity of proteins not only at the plasma membrane but also in intracellular organelles including the endoplasmic reticulum, nucleus, and mitochondria. In this review, we focus on the interactions of MaxiK channels with seven-transmembrane G protein-coupled receptors and discuss information suggesting that, the channel big C-terminus may act as the nucleus of signalling molecules including kinases relevant for cell death and survival. Increasing evidence indicates that the channel is able to associate with a variety of receptors including β-adrenergic receptors, G protein-coupled estrogen receptors, acetylcholine receptors, thromboxane A2 receptors, and angiotensin II receptors, which highlights the varied functions that the channel has (or may have) not only in regulating contraction/relaxation of muscle cells or neurotransmission in the brain but also in cell metabolism, proliferation, migration, and gene expression. In line with this view, MaxiK channels have been implicated in obesity and in brain, prostate, and mammary cancers. A better understanding on the molecular mechanisms underlying or triggered by MaxiK channel abnormalities like overexpression in certain cancers may lead to new therapeutics to prevent devastating diseases.
Collapse
|
10
|
Wang M, Jiang L, Monticone RE, Lakatta EG. Proinflammation: the key to arterial aging. Trends Endocrinol Metab 2014; 25:72-9. [PMID: 24365513 PMCID: PMC3917314 DOI: 10.1016/j.tem.2013.10.002] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 09/27/2013] [Accepted: 10/02/2013] [Indexed: 01/03/2023]
Abstract
Arterial aging is the major contributing factor to increases in the incidence and prevalence of cardiovascular disease, due mainly to the presence of chronic, low-grade, 'sterile' arterial inflammation. Inflammatory signaling driven by the angiotensin II cascade perpetrates adverse age-associated arterial structural and functional remodeling. The aged artery is characterized by endothelial disruption, enhanced vascular smooth muscle cell (VMSC) migration and proliferation, extracellular matrix (ECM) deposition, elastin fracture, and matrix calcification/amyloidosis/glycation. Importantly, the molecular mechanisms of arterial aging are also relevant to the pathogenesis of hypertension and atherosclerosis. Age-associated arterial proinflammation is to some extent mutable, and interventions to suppress or delay it may have the potential to ameliorate or retard age-associated arterial diseases.
Collapse
Affiliation(s)
- Mingyi Wang
- Laboratory of Cardiovascular Science, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD 21224, USA.
| | - Liqun Jiang
- Laboratory of Cardiovascular Science, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Robert E Monticone
- Laboratory of Cardiovascular Science, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD 21224, USA.
| |
Collapse
|
11
|
Valanti E, Tsompanidis A, Sanoudou D. Pharmacogenomics in the development and characterization of atheroprotective drugs. Methods Mol Biol 2014; 1175:259-300. [PMID: 25150873 DOI: 10.1007/978-1-4939-0956-8_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Atherosclerosis is the main cause of cardiovascular disease (CVD) and can lead to stroke, myocardial infarction, and death. The clinically available atheroprotective drugs aim mainly at reducing the levels of circulating low-density lipoprotein (LDL), increasing high-density lipoprotein (HDL), and attenuating inflammation. However, the cardiovascular risk remains high, along with morbidity, mortality, and incidence of adverse drug events. Pharmacogenomics is increasingly contributing towards the characterization of existing atheroprotective drugs, the evaluation of novel ones, and the identification of promising, unexplored therapeutic targets, at the global molecular pathway level. This chapter presents highlights of pharmacogenomics investigations and discoveries that have contributed towards the elucidation of pharmacological atheroprotection, while opening the way to new therapeutic approaches.
Collapse
Affiliation(s)
- Efi Valanti
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens, 115 27, Greece
| | | | | |
Collapse
|
12
|
Rateri DL, Moorleghen JJ, Knight V, Balakrishnan A, Howatt DA, Cassis LA, Daugherty A. Depletion of endothelial or smooth muscle cell-specific angiotensin II type 1a receptors does not influence aortic aneurysms or atherosclerosis in LDL receptor deficient mice. PLoS One 2012; 7:e51483. [PMID: 23236507 PMCID: PMC3517567 DOI: 10.1371/journal.pone.0051483] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 11/06/2012] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Whole body genetic deletion of AT1a receptors in mice uniformly reduces hypercholesterolemia and angiotensin II-(AngII) induced atherosclerosis and abdominal aortic aneurysms (AAAs). However, the role of AT1a receptor stimulation of principal cell types resident in the arterial wall remains undefined. Therefore, the aim of this study was to determine whether deletion of AT1a receptors in either endothelial cells or smooth muscle cells influences the development of atherosclerosis and AAAs. METHODOLOGY/PRINCIPAL FINDINGS AT1a receptor floxed mice were developed in an LDL receptor -/- background. To generate endothelial or smooth muscle cell specific deficiency, AT1a receptor floxed mice were bred with mice expressing Cre under the control of either Tie2 or SM22, respectively. Groups of males and females were fed a saturated fat-enriched diet for 3 months to determine effects on atherosclerosis. Deletion of AT1a receptors in either endothelial or smooth muscle cells had no discernible effect on the size of atherosclerotic lesions. We also determined the effect of cell-specific AT1a receptor deficiency on atherosclerosis and AAAs using male mice fed a saturated fat-enriched diet and infused with AngII (1,000 ng/kg/min). Again, deletion of AT1a receptors in either endothelial or smooth muscle cells had no discernible effects on either AngII-induced atherosclerotic lesions or AAAs. CONCLUSIONS Although previous studies have demonstrated whole body AT1a receptor deficiency diminishes atherosclerosis and AAAs, depletion of AT1a receptors in either endothelial or smooth muscle cells did not affect either of these vascular pathologies.
Collapse
MESH Headings
- Animals
- Aortic Aneurysm, Abdominal/etiology
- Aortic Aneurysm, Abdominal/pathology
- Atherosclerosis/etiology
- Atherosclerosis/pathology
- Blood Pressure
- Chromatography, Gel
- Crosses, Genetic
- Diet, High-Fat
- Endothelial Cells/metabolism
- Female
- Genotype
- Image Processing, Computer-Assisted
- Male
- Mice
- Mice, Knockout
- Muscle, Smooth/metabolism
- Receptor, Angiotensin, Type 1/deficiency
- Receptors, LDL/genetics
Collapse
Affiliation(s)
- Debra L. Rateri
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Jessica J. Moorleghen
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Victoria Knight
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Anju Balakrishnan
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Deborah A. Howatt
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Lisa A. Cassis
- Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
13
|
Furusho Y, Miyata M, Matsuyama T, Nagai T, Li H, Akasaki Y, Hamada N, Miyauchi T, Ikeda Y, Shirasawa T, Ide K, Tei C. Novel Therapy for Atherosclerosis Using Recombinant Immunotoxin Against Folate Receptor β-Expressing Macrophages. J Am Heart Assoc 2012; 1:e003079. [PMID: 23130174 PMCID: PMC3487340 DOI: 10.1161/jaha.112.003079] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 06/19/2012] [Indexed: 12/02/2022]
Abstract
Background Folate receptor β (FRβ) is induced during macrophage activation. A recombinant immunotoxin consisting of the truncated Pseudomonas exotoxin A (PE38) conjugated to an anti-FRβ antibody (anti–FRβ-PE38) has been reported to kill activated macrophages in inflammatory diseases. To elucidate the effect of an immunotoxin targeting FRβ on atherosclerosis, we determined the presence of FRβ-expressing macrophages in atherosclerotic lesions and administered the FRβ immunotoxin in apolipoprotein E–deficient mice. Methods and Results The FRβ-expressing macrophages were observed in atherosclerotic lesions of apolipoprotein E–deficient mice. At 15 or 35 weeks of age, the apolipoprotein E–deficient mice were divided into 3 groups and were intravenously administered 0.1 mg/kg of anti–FRβ-PE38 (immunotoxin group), 0.1 mg/kg of PE38 (toxin group), or 0.1 mL of saline (control group) every 3 days, for a total of 5 times for each age group. The mice were analyzed at 21 or 41 weeks of age. Treatment with the immunotoxin resulted in 31% and 22% reductions in atherosclerotic lesions of the 21- and 41-week-old mice, respectively (P<0.05). Administration of immunotoxin reduced the numbers of FRβ- and tumor necrosis factor-α–expressing macrophages, reduced cell proliferation, and increased the number of apoptotic cells (P<0.05). Real-time polymerase chain reaction demonstrated that the expression of FRβ and tumor necrosis factor-α mRNA was significantly decreased in the immunotoxin group (P<0.05). Conclusions These results suggest that FRβ-expressing macrophages exist in the atherosclerotic lesions of apolipoprotein E–deficient mice and that FRβ immunotoxin administration reduces the progression of atherosclerotic lesions in younger and older individuals. The recombinant FRβ immunotoxin targeting activated macrophages could provide a novel therapeutic tool for atherosclerosis. (J Am Heart Assoc. 2012;1:e003079 doi: 10.1161/JAHA.112.003079.)
Collapse
Affiliation(s)
- Yuko Furusho
- Department of Cardiovascular, Respiratory, and Metabolic Medicine, Kagoshima University, Kagoshima, Japan (Y.F., M.M., Y.A., N.H., T. Miyauchi, Y.I., T.S., K.I., C.T.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Yancey PG, Ding Y, Fan D, Blakemore JL, Zhang Y, Ding L, Zhang J, Linton MF, Fazio S. Low-density lipoprotein receptor-related protein 1 prevents early atherosclerosis by limiting lesional apoptosis and inflammatory Ly-6Chigh monocytosis: evidence that the effects are not apolipoprotein E dependent. Circulation 2011; 124:454-64. [PMID: 21730304 DOI: 10.1161/circulationaha.111.032268] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND We previously demonstrated that macrophage low-density lipoprotein receptor (LDLR)-related protein 1 (LRP1) deficiency increases atherosclerosis despite antiatherogenic changes including decreased uptake of remnants and increased secretion of apolipoprotein E (apoE). Thus, our objective was to determine whether the atheroprotective effects of LRP1 require interaction with apoE, one of its ligands with multiple beneficial effects. METHODS AND RESULTS We examined atherosclerosis development in mice with specific deletion of macrophage LRP1 (apoE(-/-) MΦLRP1(-/-)) and in LDLR(-/-) mice reconstituted with apoE(-/-) MΦLRP1(-/-) bone marrow. The combined absence of apoE and LRP1 promoted atherogenesis more than did macrophage apoE deletion alone in both apoE-producing LDLR(-/-) mice (+88%) and apoE(-/-) mice (+163%). The lesions of both mouse models with apoE(-/-) LRP1(-/-) macrophages had increased macrophage content. In vitro, apoE and LRP1 additively inhibit macrophage apoptosis. Furthermore, there was excessive accumulation of apoptotic cells in lesions of both LDLR(-/-) mice (+110%) and apoE(-/-) MΦLRP1(-/-) mice (+252%). The apoptotic cell accumulation was partially due to decreased efferocytosis as the ratio of free to cell-associated apoptotic nuclei was 3.5-fold higher in lesions of apoE(-/-) MΦLRP1(-/-) versus apoE(-/-) mice. Lesion necrosis was also increased (6 fold) in apoE(-/-) MΦLRP1(-/-) versus apoE(-/-) mice. Compared with apoE(-/-) mice, the spleens of apoE(-/-) MΦLRP1(-/-) mice contained 1.6- and 2.4-fold more total and Ly6-C(high) monocytes. Finally, there were 3.6- and 2.4-fold increases in Ly6-C(high) and CC-chemokine receptor 2-positive cells in lesions of apoE(-/-) MΦLRP1(-/-) versus apoE(-/-) mice, suggesting that accumulation of apoptotic cells enhances lesion development and macrophage content by promoting the recruitment of inflammatory monocytes. CONCLUSION Low-density lipoprotein receptor protein 1 exerts antiatherogenic effects via pathways independent of apoE involving macrophage apoptosis and monocyte recruitment.
Collapse
Affiliation(s)
- Patricia G Yancey
- Atherosclerosis Research Unit, Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Avenue, Nashville, TN 37232, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Allard JB, Duan C. Comparative endocrinology of aging and longevity regulation. Front Endocrinol (Lausanne) 2011; 2:75. [PMID: 22654825 PMCID: PMC3356063 DOI: 10.3389/fendo.2011.00075] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 10/28/2011] [Indexed: 01/06/2023] Open
Abstract
Hormones regulate growth, development, metabolism, and other complex processes in multicellular animals. For many years it has been suggested that hormones may also influence the rate of the aging process. Aging is a multifactorial process that causes biological systems to break down and cease to function in adult organisms as time passes, eventually leading to death. The exact underlying causes of the aging process remain a topic for debate, and clues that may shed light on these causes are eagerly sought after. In the last two decades, gene mutations that result in delayed aging and extended longevity have been discovered, and many of the affected genes have been components of endocrine signaling pathways. In this review we summarize the current knowledge on the roles of endocrine signaling in the regulation of aging and longevity in various animals. We begin by discussing the notion that conserved systems, including endocrine signaling pathways, "regulate" the aging process. Findings from the major model organisms: worms, flies, and rodents, are then outlined. Unique lessons from studies of non-traditional models: bees, salmon, and naked mole rats, are also discussed. Finally, we summarize the endocrinology of aging in humans, including changes in hormone levels with age, and the involvement of hormones in aging-related diseases. The most well studied and widely conserved endocrine pathway that affects aging is the insulin/insulin-like growth factor system. Mutations in genes of this pathway increase the lifespan of worms, flies, and mice. Population genetic evidence also suggests this pathway's involvement in human aging. Other hormones including steroids have been linked to aging only in a subset of the models studied. Because of the value of comparative studies, it is suggested that the aging field could benefit from adoption of additional model organisms.
Collapse
Affiliation(s)
- John B. Allard
- Department of Molecular, Cellular, and Developmental Biology, University of MichiganAnn Arbor, MI, USA
| | - Cunming Duan
- Department of Molecular, Cellular, and Developmental Biology, University of MichiganAnn Arbor, MI, USA
- *Correspondence: Cunming Duan, Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Natural Science Building, Ann Arbor, MI 48109, USA. e-mail:
| |
Collapse
|
16
|
Abstract
Arterial remodeling over time is a cornerstone of normal systemic aging. The age-associated arterial structural and functional changes in the intima, the media, and the adventitia are closely linked to angiotensin II (Ang II) signaling. A growing line of evidence indicates that essential elements of Ang II signaling, which encompasses milk fat globule epidermal growth factor-8, calpain-1, transforming growth factor-β1, matrix metalloproteinase-2/9, monocyte chemoattractant protein-1, nicotinamide adenine dinucleotide phosphate-oxidase, and reactive oxygen species, are upregulated within the central arterial wall in rats, nonhuman primates, and humans during aging. In vitro studies show that the elevation of Ang II signaling induces the accumulation of collagen and advanced glycated end-products, the degradation of elastin, and the increased cell cycle disorder, invasion, and hypertrophy of endothelial and vascular smooth muscle cells. Further, in vivo studies demonstrate that increased Ang II signaling accelerates arterial aging. Conversely, attenuating Ang II signaling via an inhibition of angiotensin conversing enzyme or a blockade of AT1 activation retards age-associated arterial remodeling. This review attempts to integrate complex facts of Ang II signaling within the aged central arterial wall and may shed light on new therapeutic targets for arterial aging.
Collapse
Affiliation(s)
- Mingyi Wang
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | | | | |
Collapse
|
17
|
Groeschel M, Braam B. Connecting chronic and recurrent stress to vascular dysfunction: no relaxed role for the renin-angiotensin system. Am J Physiol Renal Physiol 2010; 300:F1-10. [PMID: 20980410 DOI: 10.1152/ajprenal.00208.2010] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The renin-angiotensin system (RAS) is classically considered to be a protective system for volume balance and is activated during states of volume depletion. Interestingly, one of the major pathways activating the system is the sympathetic nervous system, also the primary mediator of the acute stress response. When one further examines the cells mediating the immune site of the response, which is primarily an inflammatory response leading to defense at a locally injured area, these cells all express the ANG II type 1 receptor (AGTR1). Scattered throughout the literature are reports indicating that acute and chronic stress can activate renin and increase plasma levels of components of the RAS. Moreover, there are reports describing that ANG II can modulate the distribution and function of immune cells. Since the inflammatory response is also implicated to be central in the initiation and progression of vascular damage, we propose in this review that recurrent acute stress and chronic stress can induce a state with inflammation, due to ANG II-mediated activation of inflammatory cells, specifically monocytes and lymphocytes. Such a proposal would explain a lot of the observations regarding RAS components in inflammatory cells. Despite its attractiveness, substantial research in this area would be required to substantiate this hypothesis.
Collapse
Affiliation(s)
- Michael Groeschel
- Department of Physiology, University of Alberta, and University of Alberta Hospital, Department of Medicine/Division of Nephrology and Immunology, 11-132 CSB Clinical Sciences Bldg., Edmonton, Alberta, Canada T6G 2G3
| | | |
Collapse
|
18
|
Abd Alla J, Langer A, Elzahwy SS, Arman-Kalcek G, Streichert T, Quitterer U. Angiotensin-converting enzyme inhibition down-regulates the pro-atherogenic chemokine receptor 9 (CCR9)-chemokine ligand 25 (CCL25) axis. J Biol Chem 2010; 285:23496-505. [PMID: 20504763 DOI: 10.1074/jbc.m110.117481] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Many experimental and clinical studies suggest a relationship between enhanced angiotensin II release by the angiotensin-converting enzyme (ACE) and the pathophysiology of atherosclerosis. The atherosclerosis-enhancing effects of angiotensin II are complex and incompletely understood. To identify anti-atherogenic target genes, we performed microarray gene expression profiling of the aorta during atherosclerosis prevention with the ACE inhibitor, captopril. Atherosclerosis-prone apolipoprotein E (apoE)-deficient mice were used as a model to decipher susceptible genes regulated during atherosclerosis prevention with captopril. Microarray gene expression profiling and immunohistology revealed that captopril treatment for 7 months strongly decreased the recruitment of pro-atherogenic immune cells into the aorta. Captopril-mediated inhibition of plaque-infiltrating immune cells involved down-regulation of the C-C chemokine receptor 9 (CCR9). Reduced cell migration correlated with decreased numbers of aorta-resident cells expressing the CCR9-specific chemoattractant factor, chemokine ligand 25 (CCL25). The CCL25-CCR9 axis was pro-atherogenic, because inhibition of CCR9 by RNA interference in hematopoietic progenitors of apoE-deficient mice significantly retarded the development of atherosclerosis. Analysis of coronary artery biopsy specimens of patients with coronary artery atherosclerosis undergoing bypass surgery also showed strong infiltrates of CCR9-positive cells in atherosclerotic lesions. Thus, the C-C chemokine receptor, CCR9, exerts a significant role in atherosclerosis.
Collapse
Affiliation(s)
- Joshua Abd Alla
- Molecular Pharmacology Unit, Swiss Federal Institute of Technology and University of Zurich, CH-8057 Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
19
|
Daugherty A, Poduri A, Chen X, Lu H, Cassis LA. Genetic variants of the Renin Angiotensin system: effects on atherosclerosis in experimental models and humans. Curr Atheroscler Rep 2010; 12:167-73. [PMID: 20425255 PMCID: PMC2857789 DOI: 10.1007/s11883-010-0109-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The renin angiotensin system (RAS) has profound effects on atherosclerosis development in animal models, which is partially complimented by evidence in the human disease. Although angiotensin II was considered to be the principal effector of the RAS, a broader array of bioactive angiotensin peptides have been identified that have increased the scope of enzymes and receptors in the RAS. Genetic interruption of the synthesis of these peptides has not been extensively performed in experimental or human studies. A few studies demonstrate that interruption of a component of the angiotensin peptide synthesis pathway reduces experimental lesion formation. The evidence in human studies has not been consistent. Conversely, genetic manipulation of the RAS receptors has demonstrated that AT1a receptors are profoundly involved in experimental atherosclerosis. Few studies have reported links of genetic variants of angiotensin II receptors to human atherosclerotic diseases. Further genetic studies are needed to define the role of RAS in atherosclerosis.
Collapse
Affiliation(s)
- Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky, BBSRB, Room B-243, Lexington, KY 40536-0509, USA.
| | | | | | | | | |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Age-associated arterial alterations in cells, matrix, and biomolecules are the foundation for the initiation and progression of cardiovascular diseases in older persons. This review focuses on the latest advances on the intertwining of aging and disease within the arterial wall at the cell and molecular levels. RECENT FINDINGS Endothelial dysfunction, vascular smooth muscle cell (VSMC) proliferation/invasion/secretion, matrix fragmentation, collagenization and glycation are characteristics of an age-associated arterial phenotype that creates a microenvironment enriched with reactive oxygen species (ROS) for the pathogenesis of arterial disease. This niche creates an age-associated arterial secretory phenotype (AAASP), which is orchestrated by the concerted effects of numerous age-modified angiotensin II signaling molecules. Most of these biomolecular, cell, and matrix modifications that constitute the AAASP can be elicited by experimental hypertension or atherosclerosis at a younger age. The arterial AAASP also shares features of a senescence-associated secretory phenotype (SASP) identified in other mesenchymocytes, that is, fibroblasts. SUMMARY A subclinical AAASP evolves during aging. Targeting this subclinical AAASP may reduce the incidence and progression of the quintessential age-associated arterial diseases, that is, hypertension and atherosclerosis.
Collapse
|
21
|
Shirasawa T, Miyata M, Eto H, Hamada N, Akasaki Y, Miyauchi T, Furusho Y, Orihara K, Hamasaki S, Aronow BJ, Smith JD, Tei C. Deficiency of clusterin inhibits neointimal hyperplasia after vascular injury. J Atheroscler Thromb 2009; 16:772-81. [PMID: 20032585 DOI: 10.5551/jat.1917] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Increased clusterin mRNA and protein levels have been detected in various tissues undergoing stress, and we previously reported that clusterin is markedly induced in media and neointima following vascular injury. The present study therefore investigated the impact of clusterin on neointimal hyperplasia following vascular injury. METHODS AND RESULTS As compared with wild-type mice, clusterin knockout mice (clusterin-KO) demonstrated a significant decrease of the intima/media ratio 4 weeks after cuff placement. Immunohistochemical analysis of injured femoral arteries in clusterin-KO demonstrated the accumulation of p53 in nuclei of neointimal vascular smooth muscle cells (VSMCs). Moreover, VSMCs from either clusterin-KO or rat VSMCs treated with clusterin-short-interfering (si) RNA subjected to static stretch exhibited significantly increased p53 and p21, and increased G1 cell cycle arrest as indicated by flow cytometry compared with VSMCs from wild-type mice. CONCLUSION Reduced clusterin expression reduced the proliferation of VSMCs and induced G1 arrest via p53 and p21. Clusterin therefore represents a promising molecular target to limit restenosis after coronary intervention.
Collapse
Affiliation(s)
- Takahiro Shirasawa
- Department of Cardiovascular, Respiratory and Metabolic Medicine, Kagoshima University, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Current world literature. Curr Opin Lipidol 2009; 20:512-9. [PMID: 19935200 DOI: 10.1097/mol.0b013e328334096a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
23
|
Cicero AFG, Veronesi M, Prandin MG, Di Gregori V, Ambrosioni E, Borghi C. Effects of AT1 receptor and beta1 receptor blocking on blood pressure, peripheral hemodynamic and lipid profile in statin-treated hypertensive hypercholesterolemic patients. Fundam Clin Pharmacol 2009; 23:583-8. [PMID: 19555395 DOI: 10.1111/j.1472-8206.2009.00719.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Arrigo F G Cicero
- Hypertension Research Unit, Sant'Orsola-Malpighi Hospital, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy
| | | | | | | | | | | |
Collapse
|
24
|
Umemoto S. Angiotensin II type 1 (AT1) receptor deficiency halts the progression of age-related atherosclerosis in hypercholesterolemia: molecular link between the AT1 receptor and hypercholesterolemia. Hypertens Res 2008; 31:1495-7. [PMID: 18971522 DOI: 10.1291/hypres.31.1495] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|